WO2017008049A1 - Anticorps hétérotypiques spécifiques contre le rotavirus humain - Google Patents

Anticorps hétérotypiques spécifiques contre le rotavirus humain Download PDF

Info

Publication number
WO2017008049A1
WO2017008049A1 PCT/US2016/041613 US2016041613W WO2017008049A1 WO 2017008049 A1 WO2017008049 A1 WO 2017008049A1 US 2016041613 W US2016041613 W US 2016041613W WO 2017008049 A1 WO2017008049 A1 WO 2017008049A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
rotavirus
seq
human
cells
Prior art date
Application number
PCT/US2016/041613
Other languages
English (en)
Inventor
Nitya NAIR
Lisa Blum
Ningguo FENG
William H. Robinson
Harry Greenberg
Mrinmoy SANYAL
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Priority to US15/737,634 priority Critical patent/US20180155411A1/en
Publication of WO2017008049A1 publication Critical patent/WO2017008049A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/12011Reoviridae
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/12011Reoviridae
    • C12N2720/12034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • RV Human rotaviruses
  • RVs are non-enveloped dsRNA viruses characterized by a triple-layered protein capsid comprised of two surface proteins (VP4, VP7), a major inner protein (VP6), and an 1 1- segmented dsRNA genome encoding at least 12 gene products, including 6 nonstructural proteins.
  • RVs infect enterocytes of the small-intestinal villi and replicate exclusively in the cytoplasm.
  • Virus infectivity is increased by proteolytic cleavage of the trimeric spike protein, VP4, to yield its stalk (VP5*) and globular head (VP8*) subunits. These cleavage products remain non-covalently associated on the mature virion surface.
  • VP8* mediates attachment to host cell glycoconjugates, while VP5* facilitates membrane penetration.
  • VP7 is the main glycoprotein constituent of the outer capsid of the mature virion.
  • RVs have vast serotypic diversity due to independent segregation of VP4 and VP7 by gene reassortment and antigenic differences in these proteins that provide the basis for strain classification into G (VP7) and P (VP4) serotypes.
  • G VP7
  • P VP4 serotype
  • a total of 15 G serotypes and 22 P genotypes have been described.
  • G1 , G2, G3, G4 and G9 as the G serotype and P[4], P[6] and P[8] as the P genotype
  • at least 10 G and 10 P types have been reported on human RVs.
  • an increasing number of human RVs with unusual G or P types and rare combinations of G and P types have been reported worldwide.
  • VP4 and VP7 are the only targets of in vitro neutralization and that feeding neutralizing Abs to either protein protects mice from homotypic and/or heterotypic challenge.
  • the atomic and related antigenic structures of VP4 and VP7 have been elucidated; both proteins contain conformationally-dependent regions that stimulate homotypic (serotype specific) as well as heterotypic (serotype cross-reactive) immunity.
  • the characterization of heterotypic versus homotypic interactions of VP4 and VP7 neutralizing human Abs with the virion have not been fully studied.
  • RV-specific Ab clones generated from a bone marrow-derived phage display library.
  • three neutralizing human mAbs were identified; two VP4 mAbs isolated had heterotypic specificities whereas the single VP7 mAb had only homotypic specificity.
  • a significant challenge for vaccine development is defining conserved epitopes that are capable of eliciting cross-reactive protective antibodies in this highly diverse virus.
  • Treatment of rotavirus and the development of vaccines that broadly protect against highly diverse rotavirus serotypes are of interest in the field, particularly due to the fact that lowered protective immunity to current, licensed vaccine formulations is low in regions of the world with the highest proportion or morbidity and mortality attributed to rotavirus.
  • the present invention addresses this issue.
  • mAbs Human recombinant, neutralizing monoclonal antibodies (mAbs) specific to rotavirus protein epitopes.
  • the provided antibodies were generated by cloning natively paired heavy (IgH) and light (IgL) chain antibody (Ab) genes derived from effector B cells in the small intestinal mucosa of RV-experienced adults.
  • the antibodies have heterotypic (serotype cross-reactive) neutralizing capacity against two, three or more RV strains.
  • the antibodies have homotypic (serotype specific) neutralizing activity.
  • the antibody is specific for a VP7 epitope.
  • the antibody is specific for a VP4 epitope, including the VP5* cleavage product of VP4.
  • Exemplary antibody seqyences are provided herein.
  • mAbs are useful in defining epitopes that stimulate homotypic versus heterotypic protective immunity in humans, and in the rational design of more effective RV vaccines, for example vaccines that include epitopes that stimulate heterotypic immunity against serotypically distinct RV strains circulating worldwide. These antibodies are also therapeutically useful.
  • the antibodies provided herein include VP4 specific mAbs, including without limitation antibodies specific for the VP5* cleavage product, that neutralyze RVs with diverse serotypes, including G x P6, G xP8, G x P4 and Gx P3.
  • the native antibodies are typically of an IgA isotype; in some embodiments the antibody is provided as an antibody of other than IgA isotype, e.g. lgG1 , lgG2a, lgG2b, lgG3, lgG4; as a single chain antibody; in combination with an engineered Fc region, and the like.
  • the antibody may be labeled with a detectable label, immobilized on a solid phase and/or conjugated with a heterologous compound.
  • the antibody or a cocktail of antibodies may be provided as a pharmaceutical formulation
  • Embodiments of the invention include isolated antibodies and derivatives and fragments thereof, pharmaceutical formulations comprising one or more of the human anti-rotavirus monoclonal antibodies; and cell lines that produce these monoclonal antibodies. Also provided are CDR amino acid sequences that confer the binding specificity of these monoclonal antibodies. These sequences and the cognate epitopes to which the monoclonal antibodies of the invention bind can be used to identify other antibodies that specifically bind and neutralize rotavirus; including without limitation epitopes of VP5*, and immunotherapeutic methods for prevention of disease associated with RV.
  • An advantage of the monoclonal antibodies of the invention derives from the fact that they are encoded by a human polynucleotide sequence.
  • the human anti-rotavirus antibody may have a heavy chain variable region comprising the amino acid sequence of CDR1 and/or CDR2 and/or CDR3 of the provided monoclonal antibodies as provided herein; and/or a light chain variable region comprising the amino acid sequence of CDR1 and/or CDR2 and/or CDR3 of the provided human monoclonal human antibodies as provided herein.
  • the antibody comprises an amino acid sequence variant of one or more of the CDRs of the provided human antibodies, which variant comprises one or more amino acid insertion(s) within or adjacent to a CDR residue and/or deletion(s) within or adjacent to a CDR residue and/or substitution(s) of CDR residue(s) (with substitution(s) being the preferred type of amino acid alteration for generating such variants).
  • Such variants will normally having a high binding affinity for rotavirus VP4 or VP7.
  • the invention provides a method for determining the presence of a specific serotype of human rotavirus virus exposing a sample suspected of containing the rotavirus virus to the anti-rotavirus antibody and determining binding of the antibody to the sample. While human VP6-specific mAbs have been identified in the past that would serve to identify the presence of rotavirus, VP4- and VP7-specific mAbs that react with specific serotypes would allow for diagnosis of the specific infecting human strain.
  • the invention further provides: isolated nucleic acid encoding the antibodies and variants; a vector comprising that nucleic acid, optionally operably linked to control sequences recognized by a host cell transformed with the vector; a host cell comprising that vector; a process for producing the antibody comprising culturing the host cell so that the nucleic acid is expressed and, optionally, recovering the antibody from the host cell culture (e.g. from the host cell culture medium).
  • the invention also provides a composition comprising one or more of the human anti-rotavirus antibodies and a pharmaceutically acceptable carrier or diluent. This composition for therapeutic use is sterile and may be lyophilized, e.g. being provided as a prepack in a unit dose with diluent and delivery device, e.g. inhaler, syringe, etc.
  • a basis for heterotypic neutralizing reactivity to RV in humans at the individual immunoglobulin (Ig) molecule level is identified.
  • a method of defining such activity comprising the steps of sorting single cells of intestinal RV-specific lgA + antibody secreting cells, by contacting the cells with triple-layered RV particles conjugated to a detectable label, e.g. a fluorochrome suitable for sorting by flow cytometry.
  • the immunoglobulin coding polynucleotides from such sorted cells are sequenced with an identifying barcode.
  • the antibodies thus identified by sequences are tested for activity in RV neutralization in vitro against two or more different RV serotypes, where antibodies that neutralize multiple serotypes are defined as heterotypic antibodies.
  • the methods are useful in providing detailed analysis of thre ability of an immunogen, e.g. a vaccine, to elicit a protective heterotypic response.
  • Humans can circumvent the serotypic diversity of naturally circulating RV strains by expressing individual VP4 epitope-specific Ig molecules that mediate heterotypic neutralization. Characterization of the structural targets of these mAbs, and determination of the extent to which they arise following primary RV infection of children provide the basis for designing more effective RV vaccines.
  • Antigenic compositions comprise all or a portion of a rotavirus protein in which specific highly immunodominant residues are masked or deleted, so as to generate an immune response to residues that are less immunodominant, but which are essential for virus function and therefore are less likely to be altered in virus escape mutation and selection.
  • antigenic compositions providing epitopes for heterotypic neutralizing antibodies are provided, which can be formulated alone or in combination with conventional vaccines.
  • Antigens may comprise, without limitation, VP5* proteins, alone or in combination with an adjuvant. These antigens find use in screening assays, generation of monoclonal antibodies, and in vaccines.
  • Such formulations may comprise, without limitation, live attenuated formulation containing known heterotypic neutralizing epitopes (and excluding known homotypic neutralizing epitopes); and/or epitope immunogens with known heterotypic neutralizing epitopes or overlapping neutralizing epitopes.
  • novel vaccines/immunogens could be used in combination with current formulations, for example in a prime boost strategy to enhance immunity in children and infants who do not respond to the current, licensed vaccines or formulations alone.
  • the formulations of the invention may find particular benefit in providing improved protective immunity in regions of the world with the highest RV disease burden and lowest vaccine efficacy observed in several clinical trials of the current licensed RV vaccines.
  • a modified rotavirus VP4, including a VP5* fragment, or VP7 polypeptide is provided, which provides for enhanced heterotypic immune responsiveness
  • a polynucleotide encoding such a modified rotavirus polypeptide is provided.
  • the polypeptide and/or the nucleic acid can be used in the formulation of a vaccine, e.g. a virus-like particle, a recombinant protein vaccine which can be formulated with an adjuvant, a vector vaccine, and the like.
  • a vaccine formulation comprising a polypeptide or a polynucleotide of the invention is provided.
  • FIG. 1A-1 E Identification of RV-specific antibody secreting B cells by flow cytometry using triple layered particles (TLP)-Cy5.
  • RV TLPs CDC-9 strain, G1 , P[8]
  • Cy5 the structural integrity of the TLPs-Cy5 determined by electron microscopy TLP- Cy5 specifically stained G1- and P8- specific murine hybridomas but did not stain G3- and PS- specific hybridomas by FACS.
  • B TLP-Cy5 binding to G1- and P8- specific hybridomas was reduced by blocking with unlabeled TLPs.
  • TLP-Cy5 bound VP6-specific hybridoma ; Treatment of TLP-Cy5 with 5 mM EDTA increased the proportion of VP6-specific hybridoma cells that stained positive by FACS compared to untreated TLP-Cy5.
  • C Shown are represented histogram overlays and the mean fluorescence intensity + SD from two independent experiments. At least 100, 000 events were acquired per sample.
  • D Human intestinal ASCs were identified by FACS based on single, live, CD3/14/16 " CD20 lo/" CD27 hi CD38 hi surface phenotype. TLP-Cy5-binding B cells were gated based on unstained cells.
  • Blocking with unlabeled TLPs reduced TLP-Cy5-specific staining on intestinal ASCs. Shown are FACS plots from a representative donor and the mean frequency of intestinal TLP + ASCs + SD from repeated experiments on two donors. At least 200,000 events were acquired per sample; and (E) the mean frequency of intestinal TLP+ ASCs + SD from repeated experiments on two donors. At least 200,000 events were acquired per sample. P values were obtained using oneway ANOVA. *, P ⁇ 0.05; **, P ⁇ 0.01 ; ****, P ⁇ 0.0001 .
  • FIG. 2A-2C Identification and frequency of TLP-binding human intestinal ASCs at steady-state in adults donors.
  • A Gating strategy used to identify CDC-9 TLP-binding intestinal ASCs derived from proximal jejunum tissue resections of adult donors. Live, single cells were gated based on CD3/14/16 " CD20 lo/" CD27 hi CD38 hi lgA + surface expression. Shown are contour plots from a representative donor. At least 200,000 events were acquired per sample.
  • (B) The frequency of lgA+ and IgA- ASCs as a proportion of total intestinal ASCs (left) and the frequency of TLP-binding lgA+ and IgA- ASCs as a proportion of total lgA+ and IgA- ASCs (right) , as determined by FACS are shown.
  • (C) The frequency of lgA+ ASCs as a proportion of total intestinal B cells (left) and the frequency of DLP-binding lgA+ ASCs as a proportion of total lgA+ ASCs (right) as determined by ELISPOT. Symbols represent the frequencies of individual donors as shown in the legend to the right. Red lines represent the median frequencies from five donors. P values were obtained using the unpaired t test. *, P ⁇ 0.05.
  • FIG. 3 Phylogenetic tree of the RV TLP-reactive lgA+ ASC intestinal Ab repertoire. Combined heavy and light chain dendrograms of the Ab repertoires of TLP-binding intestinal lgA+ ASCs from five donors. The subject ID and the total number of paired Ab sequences used to generate each phylogenetic tree are shown in the center. Each peripheral node depicts a sequenced VH and VL region derived from a single cell. Colors indicate VH gene families as indicated in the legend to the right and red lines indicate clonal families. Ig V gene sequences that were selected for cloning and expression of recombinant mAbs are numbered. Stars denote Abs that bound RV proteins, circled red stars denote neutralizing Abs, and squares denote Abs that did not bind RV.
  • FIGS 4A-4E Molecular characteristics of paired IgH and IgL immunoglobulin genes expressed by individual TLP-reactive intestinal IgA+ ASCs.
  • A Heatmap representation of VH- VL combinations that occurred in more than one donor among lgA+ ASC Ab sequences. Colors indicate the sequence-normalized number of Abs per combination as shown in the scale below the heatmap.
  • B The frequency of replacement (black) and silent (white) mutations in FWRs and CDRs of the 821 lgA+ ASC gene sequences analyzed.
  • FIGS 5A-5C Recombinant human mAbs can mediate heterotypic as well as homotypic protection from RV-induced diarrheal disease.
  • the terms “neutralizes rotavirus,” “inhibits rotavirus,” and “blocks rotavirus” are used interchangeably to refer to the ability of an antibody of the invention to prevent rotavirus from infecting a given cell.
  • the term "effective dose” or “effective dosage” is defined as an amount sufficient to achieve or at least partially achieve the desired effect.
  • therapeutically effective dose is defined as an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient's own immune system.
  • a heterotypic antibody is capable of neutralizing two, three, four or more different rotavirus serotypes.
  • a “homotypic” antibody neutralizes specifically a single serotype, particularly the serotype used as an immunogen.
  • rotavirus protein includes without limitation the proteins, particularly VP4, VP7 and fragments thereof, of known serotypes that infect humans, e.g. as described in Hemming and Vesikari (2013) Infect Genet Evol. Oct; 19:51 -8; Lahon and Chitambar (201 1) Asian Pac J Trap Med. Nov;4(1 1):846-9; Arora et al. (201 1) Asian Pac J Trap Med. Jul;4(7):541 -6; Aung et al. (2009) J Med Virol. 2009 Nov;81 (1 1): 1968-74; Yoder et al. (2009) J Virol. 2009 Nov;83(21): 1 1372-7, each herein specifically incorporated by reference.
  • VP5* epitopes are shown herein to be associated with heterotypic antibody responses. Proteolytic cleavage of the VP4 outer capsid spike protein into VP8* and VP5* proteins is required for rotavirus infectivity and for rotavirus-induced membrane permeability.
  • the cleavage site may be at about amino acid 247-248 of VP4, thus the VP5* fragment may comprise from about residue 247 to about residue 775.
  • a recombinant VP5* fragment has a trimeric, folded-back structure.
  • VP5* forms the spike body and foot and is thought to mediate membrane penetration. The head and body domains form an asymmetrical dyad that protrudes from the VP7 shell.
  • composition/method/kit By “comprising” it is meant that the recited elements are required in the composition/method/kit, but other elements may be included to form the composition/method/kit etc. within the scope of the claim.
  • treatment covers any treatment of a disease in a mammal, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; or (c) relieving the disease, i.e., causing regression of the disease.
  • the therapeutic agent may be administered before, during or after the onset of disease or injury.
  • the treatment of ongoing disease, where the treatment stabilizes or reduces the undesirable clinical symptoms of the patient, is of particular interest. Such treatment is desirably performed prior to complete loss of function in the affected tissues.
  • the subject therapy may be administered during the symptomatic stage of the disease, and in some cases after the symptomatic stage of the disease.
  • Polypeptide and “protein” as used interchangeably herein, can encompass peptides and oligopeptides. Where “polypeptide” is recited herein to refer to an amino acid sequence of a naturally-occurring protein molecule, "polypeptide” and like terms are not necessarily limited to the amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule, but instead can encompass biologically active variants or fragments, including polypeptides having substantial sequence similarity or sequence identify relative to the amino acid sequences provided herein. In general, fragments or variants retain a biological activity of the parent polypeptide from which their sequence is derived.
  • polypeptide refers to an amino acid sequence of a recombinant or non-recombinant polypeptide having an amino acid sequence of i) a native polypeptide, ii) a biologically active fragment of an polypeptide, or iii) a biologically active variant of an polypeptide.
  • Polypeptides suitable for use can be obtained from any species, e.g. , mammalian or non-mammalian (e.g. , reptiles, amphibians, avian (e.g.
  • polypeptides comprising a sequence of a human polypeptide are of particular interest.
  • the term "derived from” indicates molecule that is obtained directly from the indicated source (e.g., when a protein directly purified from a cell, the protein is “derived from” the cell) or information is obtained from the source, e.g. nucleotide or amino acid sequence, from which the molecule can be synthesized from materials other than the source of information.
  • isolated indicates that the recited material (e.g, polypeptide, nucleic acid, etc.) is substantially separated from, or enriched relative to, other materials with which it occurs in nature (e.g., in a cell).
  • a material e.g., polypeptide, nucleic acid, etc.
  • a material that is isolated constitutes at least about 0.1 %, at least about 0.5%, at least about 1 % or at least about 5% by weight of the total material of the same type (e.g., total protein, total nucleic acid) in a given sample.
  • subject and patient are used interchangeably herein to mean a member or members of any mammalian or non-mammalian species that may have a need for the pharmaceutical methods, compositions and treatments described herein.
  • Subjects and patients thus include, without limitation, primate (including humans), canine, feline, ungulate (e.g., equine, bovine, swine (e.g., pig)), avian, and other subjects.
  • Humans and non-human animals having commercial importance are of particular interest.
  • subject and patient refer to a subject or patient susceptible to infection by a Flaviviridae virus, particularly rotavirus.
  • mammalian means a member or members of any mammalian species, and includes, by way of example, canines; felines; equines; bovines; ovines; rodentia, etc. and primates, particularly humans.
  • Non-human animal models, particularly mammals, e.g. primate, murine, lagomorpha, etc. may be used for experimental investigations.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the novel unit dosage forms depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • a "pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” and “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, and adjuvant that are useful in preparing a pharmaceutical composition that are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, and adjuvant that are acceptable for veterinary use as well as human pharmaceutical use.
  • “A pharmaceutically acceptable excipient, diluent, carrier and adjuvant” as used in the specification and claims includes both one and more than one such excipient, diluent, carrier, and adjuvant.
  • a "pharmaceutical composition” is meant to encompass a composition suitable for administration to a subject, such as a mammal, especially a human.
  • a “pharmaceutical composition” is sterile, and is usually free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade).
  • Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal and the like.
  • epitopic determinants means any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • conjugate as described and claimed herein is defined as a heterogeneous molecule formed by the covalent attachment of one or more polypeptide fragment(s) to one or more polymer molecule(s), wherein the heterogeneous molecule is water soluble, i.e. soluble in physiological fluids such as blood, and wherein the heterogeneous molecule is free of any structured aggregate.
  • a conjugate of interest is PEG.
  • structured aggregate refers to (1) any aggregate of molecules in aqueous solution having a spheroid or spheroid shell structure, such that the heterogeneous molecule is not in a micelle or other emulsion structure, and is not anchored to a lipid bilayer, vesicle or liposome; and (2) any aggregate of molecules in solid or insolubilized form, such as a chromatography bead matrix, that does not release the heterogeneous molecule into solution upon contact with an aqueous phase.
  • conjugate encompasses the aforementioned heterogeneous molecule in a precipitate, sediment, bioerodible matrix or other solid capable of releasing the heterogeneous molecule into aqueous solution upon hydration of the solid.
  • label when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the antibody.
  • the label may itself be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • solid phase is meant a non-aqueous matrix to which the antibody of the present invention can adhere.
  • solid phases encompassed herein include those formed partially or entirely of glass (e.g. controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones.
  • the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g. an affinity chromatography column). This term also includes a discontinuous solid phase of discrete particles, such as those described in U.S. Pat. No. 4,275,149.
  • vaccine as used herein, is meant a composition; a formulation comprising a modified polypeptide of the invention; a virus or virus-like particle comprising a modified polypeptide of the invention complex; or a DNA encoding a modified polypeptide of the invention complex, which, when administered to a subject, induces cellular or humoral immune responses as described herein.
  • Some embodiments of the invention provide a method of stimulating an immune response in a mammal, which can be a human or a preclinical model for human disease, e.g. mouse, ape, monkey etc.
  • Stimulating an immune response includes, but is not limited to, inducing a therapeutic or prophylactic effect that is mediated by the immune system of the mammal. More specifically, stimulating an immune response in the context of the invention refers to eliciting cellular or humoral immune responses, thereby inducing downstream effects such as production of antibodies, antibody heavy chain class switching, maturation of APCs, and stimulation of cytolytic T cells, T helper cells and both T and B memory cells.
  • vaccine compositions are suitably formulated to be compatible with the intended route of administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as
  • the pH of the composition can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • Systemic administration of the composition is also suitably accomplished by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • Vaccine compositions may include an aqueous medium, pharmaceutically acceptable inert excipient such as lactose, starch, calcium carbonate, and sodium citrate.
  • Vaccine compositions may also include an adjuvant, for example Freud's adjuvant.
  • Vaccines may be administered alone or in combination with a physiologically acceptable vehicle that is suitable for administration to humans.
  • Vaccines may be delivered orally, parenterally, intramuscularly, intranasally or intravenously. Oral delivery may encompass, for example, adding the compositions to the feed or drink of the mammals. Factors bearing on the vaccine dosage include, for example, the weight and age of the mammal.
  • Compositions for parenteral or intravenous delivery may also include emulsifying or suspending agents or diluents to control the delivery and dose amount of the vaccine.
  • Modified polypeptides of the invention and polynucleotides that encode such modified polypeptides can be used in various rotavirus vaccine formulations known in the art, as a substitution for the wild-type rotavirus sequence.
  • Polypeptides can be fragmented to generate a peptide vaccine, e.g. administered with poly-L-arginine, can be formulated as a vaccine.
  • Polynucleotides encoding modified polypeptides can be administered in virus form, e.g. modified rotavirus, plasmid form, in a virus genome, including adenovirus, alphaviruses, canary pox, ovine atadenovirus and semliki-like viral particles.
  • Antibodies also referred to as immunoglobulins, conventionally comprise at least one heavy chain and one light, where the amino terminal domain of the heavy and light chains is variable in sequence, hence is commonly referred to as a variable region domain, or a variable heavy (VH) or variable light (VH) domain.
  • VH variable heavy
  • VH variable light
  • the two domains conventionally associate to form a specific binding region, although a variety of non-natural configurations of antibodies are known and used in the art.
  • a "functional” or “biologically active” antibody or antigen-binding molecule is one capable of exerting one or more of its natural activities in structural, regulatory, biochemical or biophysical events.
  • a functional antibody or other binding molecule may have the ability to specifically bind an antigen and the binding may in turn elicit or alter a cellular or molecular event such as signaling transduction or enzymatic activity.
  • a functional antibody or other binding molecule may neutralize a virus particle. The capability of an antibody or other binding molecule to exert one or more of its natural activities depends on several factors, including proper folding and assembly of the polypeptide chains.
  • antibody herein is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, monomers, dimers, multimers, multispecific antibodies (e.g. , bispecific antibodies) , single chain Fv, nanobodies, etc. , and also include antibody fragments, so long as they exhibit the desired biological activity (Miller et al (2003) Jour, of Immunology 170:4854-4861 ).
  • Antibodies may be murine, human, humanized, chimeric, or derived from other species.
  • the term antibody may reference a full-length heavy chain, a full length light chain, an intact immunoglobulin molecule; or an immunologically active portion of any of these polypeptides, i.e. , a polypeptide that comprises an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • the immunoglobulin disclosed herein can be of any type (e.g. , IgG, IgE, IgM , IgD, and IgA), class (e.g.
  • the antibody is other than a full length IgA antibody. In one aspect, the antibody is of largely human origin.
  • variable refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs).
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a beta-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al (1991 ) Sequences of Proteins of I mmunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.) .
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC).
  • ADCC antibody dependent cellular cytotoxicity
  • the hypervariable region may comprise amino acid residues from a "complementarity determining region” or "CDR”, and/or those residues from a “hypervariable loop".
  • “Framework Region” or "FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • Variable regions of interest include at least one CDR sequence from the variable regions provided herein, usually at least 2 CDR sequences, and more usually 3 CDR sequences, exemplary CDR designations are shown herein, however one of skill in the art will understand that a number of definitions of the CDRs are commonly in use, including the Kabat definition (see “Zhao et al. A germline knowledge based computational approach for determining antibody complementarity determining regions.” Mol Immunol. 2010;47:694-700), which is based on sequence variability and is the most commonly used. The Chothia definition is based on the location of the structural loop regions (Chothia et al. "Conformations of immunoglobulin hypervariable regions.” Nature.
  • CDR definitions of interest include, without limitation, those disclosed by Honegger, "Yet another numbering scheme for immunoglobulin variable domains: an automatic modeling and analysis tool.” J Mol Biol. 2001 ;309:657-670; Ofran et al. "Automated identification of complementarity determining regions (CDRs) reveals peculiar characteristics of CDRs and B cell epitopes.” J Immunol. 2008; 181 :6230-6235; Almagro “Identification of differences in the specificity- determining residues of antibodies that recognize antigens of different size: implications for the rational design of antibody repertoires.” J Mol Recognit. 2004; 17: 132-143; and Padlanet al. "Identification of specificity-determining residues in antibodies.” Faseb J. 1995;9: 133-139., each of which is herein specifically incorporated by reference.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
  • the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81 :6851-6855).
  • Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc) and human constant region sequences.
  • an "intact antibody chain” as used herein is one comprising a full length variable region and a full length constant region.
  • An intact “conventional” antibody comprises an intact light chain and an intact heavy chain, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1 , hinge, CH2 and CH3 for secreted IgG.
  • CL light chain constant domain
  • Other isotypes, such as IgM or IgA may have different CH domains.
  • the constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variants thereof.
  • the intact antibody may have one or more "effector functions" which refer to those biological activities attributable to the Fc constant region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody.
  • effector functions include C1 q binding; complement dependent cytotoxicity; Fc receptor binding; antibody- dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors.
  • Constant region variants include those that alter the effector profile, binding to Fc receptors, and the like.
  • immunoglobulin antibodies can be assigned to different "classes.” There are five major classes of intact immunoglobulin antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., lgG1 , lgG2, lgG3, lgG4, IgA, and lgA2.
  • the heavy- chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • Ig forms include hinge-modifications or hingeless forms (Roux et al (1998) J. Immunol. 161 :4083-4090; Lund et al (2000) Eur. J. Biochem. 267:7246-7256; US 2005/0048572; US 2004/0229310).
  • the light chains of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called ⁇ and ⁇ , based on the amino acid sequences of their constant domains.
  • a "functional Fc region” possesses an "effector function" of a native-sequence Fc region.
  • exemplary effector functions include C1 q binding; CDC; Fc-receptor binding; ADCC; ADCP; down-regulation of cell-surface receptors (e.g., B-cell receptor), etc.
  • Such effector functions generally require the Fc region to be interact with a receptor, e.g. the FcyRI; FcyRIIA; FcvRIIBI ; FcyRIIB2; FcyRIIIA; FcyRIIIB receptors, and the law affinity FcRn receptor; and can be assessed using various assays as disclosed, for example, in definitions herein.
  • a "dead" Fc is one that has been mutagenized to retain activity with respect to, for example, prolonging serum half-life, but which does not activate a high affinity Fc receptor.
  • a "native-sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • Native-sequence human Fc regions include a native-sequence human lgG1 Fc region (non-A and A allotypes); native-sequence human lgG2 Fc region; native-sequence human lgG3 Fc region; and native-sequence human lgG4 Fc region, as well as naturally occurring variants thereof.
  • a “variant Fc region” comprises an amino acid sequence that differs from that of a native-sequence Fc region by virtue of at least one amino acid modification, preferably one or more amino acid substitution(s).
  • the variant Fc region has at least one amino acid substitution compared to a native-sequence Fc region or to the Fc region of a parent polypeptide, e.g., from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native-sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% homology with a native-sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% homology therewith, more preferably at least about 95% homology therewith.
  • Fv is the minimum antibody fragment, which contains a complete antigen-recognition and antigen-binding site.
  • the CD3 binding antibodies of the invention comprise a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association; however additional antibodies, e.g. for use in a multi-specific configuration, may comprise a VH in the absence of a VL sequence. Even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although the affinity may be lower than that of two domain binding site.
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain.
  • Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear at least one free thiol group.
  • F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • single chain antibody means a single polypeptide chain containing one or more antigen binding domains that bind an epitope of an antigen, where such domains are derived from or have sequence identity with the variable region of an antibody heavy or light chain.
  • Parts of such variable region may be encoded by V H or V L gene segments, D and J H gene segments, or J L gene segments.
  • the variable region may be encoded by rearranged V H DJ H , V L DJ H , V H JL, or V L J L gene segments.
  • V-, D- and J-gene segments may be derived from humans and various animals including birds, fish, sharks, mammals, rodents, non- human primates, camels, lamas, rabbits and the like.
  • an "isolated" antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the antibody will be purified (1) to greater than 75% by weight of antibody as determined by the Lowry method, and most preferably more than 80%, 90% or 99% by weight, or (2) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • label when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the antibody.
  • the label may itself be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • compositions and methods are provided relating to human anti-rotavirus monoclonal antibodies.
  • the antibodies of the invention bind to and neutralize rotavirus virus across multiple genotypes.
  • Embodiments of the invention include isolated antibodies and derivatives and fragments thereof, pharmaceutical formulations comprising one or more of the human anti- rotavirus monoclonal antibodies; cell lines that produce these monoclonal antibodies.
  • the present invention is directed to combinatorially derived human monoclonal antibodies which are specifically reactive with and neutralize rotavirus, and cell lines which produce such antibodies.
  • Variable regions of exemplary antibodies are provided, e.g. SEQ ID NO: 19-36 provide protein sequences of antibodies, which may be paired, as intact variables regions or as a set of CDR sequences derived therefrom, as SEQ ID NO: 19 and 20; SEQ ID NO:21 and 22; SEQ ID NO:23 and 24; SEQ ID NO:25 and 26; SEQ ID NO:27 and 28; SEQ ID NO:29 and 30; SEQ ID NO:31 and 32; SEQ ID NO:33 and 34; and SEQ ID NO:35 and 36.
  • Combinations of particular interest include those antibodies shown to have heterotypic neutralization activity, i.e. mAb ID nos. 2, 30, 41 , 47, 49 and 57, which correspond to the combinations as intact variables regions or as a set of CDR sequences derived therefrom SEQ ID NO: 19 and 20; SEQ ID NO:23 and 24; SEQ ID NO:27 and 28; SEQ ID NO:31 and 32; SEQ ID NO:33 and 34; and SEQ ID NO:35 and 36.
  • heterotypic neutralization activity i.e. mAb ID nos. 2, 30, 41 , 47, 49 and 57
  • Antibodies of interest include these provided combinations, as well as fusions of the variable regions to appropriate constant regions or fragments of constant regions, e.g. to generate F(ab)' antibodies.
  • Variable regions of interest include at least one CDR sequence, where a CDR may be 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 or more amino acids.
  • antibodies of interest include a pair of variable regions.
  • one antibody chain can comprise the set of CDR sequences from a heavy chain. Such an antibody chain may be combined with an antibody chain comprising the set of CDR sequences from a light chain.
  • Certain antibodies of the invention bind to rotavirus VP4, including the VP5* fragment, or VP7 proteins of different rotavirus serotypes.
  • One or more residues of a CDR may be altered to modify binding to achieve a more favored on-rate of binding, a more favored off-rate of binding, or both, such that an optimized binding constant is achieved.
  • Affinity maturation techniques are well known in the art and can be used to alter the CDR region(s), followed by screening of the resultant binding molecules for the desired change in binding.
  • modifications can also be made within one or more of the framework regions, FRI, FR2, FR3 and FR4, of the heavy and/or the light chain variable regions of a human antibody, so long as these modifications do not eliminate the binding affinity of the human antibody.
  • the framework regions of human antibodies are usually substantially identical, and more usually, identical to the framework regions of the human germline sequences from which they were derived.
  • many of the amino acids in the framework region make little or no direct contribution to the specificity or affinity of an antibody.
  • many individual conservative substitutions of framework residues can be tolerated without appreciable change of the specificity or affinity of the resulting human immunoglobulin.
  • the variable framework region of the human antibody shares at least 85% sequence identity to a human germline variable framework region sequence or consensus of such sequences.
  • the variable framework region of the human antibody shares at least 90%, 95%, 96%, 97%, 98% or 99% sequence identity to a human germline variable framework region sequence or consensus of such sequences.
  • a monoclonal antibody may be selected for its retention of other functional properties of antibodies of the invention, such as binding to multiple serotypes of rotavirus and/or binding with an ultra-high affinity such as, for example, a K D of 10 9 M or lower.
  • a polypeptide of interest has a contiguous sequence of at least about 10 amino acids as set forth in any one of sequences provided herein, at least about 15 amino acids, at least about 20 amino acids, at least about 25 amino acids, at least about 30 amino acids, up to the complete provided variable region.
  • Polypeptides of interest also include variable regions sequences that differ by up to one, up to two, up to 3, up to 4, up to 5, up to 6 or more amino acids as compared to the amino acids sequence set forth in any one of sequences provided herein.
  • a polypeptide of interest is at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 99% identical to the amino acid sequence set forth in any one of sequences provided herein.
  • the isolation of cells producing monoclonal antibodies of the invention can be accomplished using routine screening techniques, which permit determination of the elementary reaction pattern of the monoclonal antibody of interest.
  • routine screening techniques which permit determination of the elementary reaction pattern of the monoclonal antibody of interest.
  • a human monoclonal antibody has the same specificity as a human monoclonal antibody of the invention by ascertaining whether the former prevents the latter from binding to or neutralizing rotavirus, including without limitation an ability to neutralize an rotavirus virus of multiple serotypes. If the human monoclonal antibody being tested competes with the human monoclonal antibody of the invention, as shown by a decrease in binding by the human monoclonal antibody of the invention, then the two monoclonal antibodies bind to the same, or a closely related, epitope.
  • Still another way to determine whether a human monoclonal antibody has the specificity of a human monoclonal antibody of the invention is to pre-incubate the human monoclonal antibody of the invention with rotavirus with which it is normally reactive, and then add the human monoclonal antibody being tested to determine if the human monoclonal antibody being tested is inhibited in its ability to bind rotavirus. If the human monoclonal antibody being tested is inhibited then, in all likelihood, it has the same, or functionally equivalent, epitopic specificity as the monoclonal antibody of the invention. Screening of human monoclonal antibodies of the invention can be also carried out utilizing rotavirus and determining whether the monoclonal antibody neutralizes rotavirus.
  • single chain antibodies can be constructed according to the method of U.S. Pat. No. 4,946,778 to Ladner et al, which is incorporated herein by reference in its entirety. Single chain antibodies comprise the variable regions of the light and heavy chains joined by a flexible linker moiety. Yet smaller is the antibody fragment known as the single domain antibody, which comprises an isolate VH single domain. Techniques for obtaining a single domain antibody with at least some of the binding specificity of the intact antibody from which they are derived are known in the art.
  • H single domain antibody antibody heavy chain variable region
  • the invention includes methods of treating an rotavirus-mediated disease in a subject by administering to the subject an isolated human monoclonal antibody or antigen binding portion thereof as described herein (i.e., that specifically binds to rotavirus), or a cocktail of such antibodies, in an amount effective to inhibit rotavirus disease, e.g., rotavirus-mediated symptoms or morbidity.
  • rotavirus disease e.g., rotavirus-mediated symptoms or morbidity.
  • diseases may include various conditions associated with rotavirus infection such as severe dehydrating diarrhea.
  • Treatment may include the use of the monoclonal antibodies of the invention as a single agent, or as an agent in combination with rehydration therapy, drugs, additional antibodies, vaccines, and the like.
  • Subjects suspected of having an rotavirus infection can be screened prior to therapy. Further, subjects receiving therapy may be tested in order to assay the activity and efficacy of the treatment. Significant improvements in one or more parameters is indicative of efficacy. It is well within the skill of the ordinary healthcare worker (e.g., clinician) to adjust dosage regimen and dose amounts to provide for optimal benefit to the patient according to a variety of factors (e.g., patient-dependent factors such as the severity of the disease and the like, the compound administered, and the like). For example, rotavirus infection in an individual can be detected and/or monitored by the presence of rotavirus RNA in blood, and/or having anti-rotavirus antibody in their serum.
  • Subjects for whom the therapy disclosed herein is of interest include subject who are "difficult to treat" subjects due to the nature of the rotavirus infection or the nature of the individual, e.g. extreme youth or age, immunosuppression, etc.
  • Human monoclonal antibodies or portions thereof (and compositions comprising the antibodies or portions thereof) of the invention can be administered in a variety of suitable fashions, e.g., intravenously (IV), subcutaneously (SC), or, intramuscularly (IM) to the subject.
  • the antibody or antigen-binding portion thereof can be administered alone or in combination with another therapeutic agent, e.g., a second human monoclonal antibody or antigen binding portion thereof.
  • the second human monoclonal antibody or antigen binding portion thereof specifically binds to a second rotavirus isolate that differs from the isolate bound to the first antibody.
  • the antibody is administered together with another agent, for example, an antiviral agent.
  • Antiviral agents includes pegylated interferon a, ribivarin, etc.
  • the antibody is administered together with a polyclonal gamma-globulin (e.g., human gammaglobulin).
  • the antibody is administered before, after, or contemporaneously with a rotavirus vaccine.
  • the human monoclonal antibodies of the invention can be used in vitro and in vivo to detect or monitor the course of rotavirus disease.
  • detect or monitor the course of rotavirus disease For example, by measuring the increase or decrease in the number of cells infected with rotavirus or changes in the concentration of rotavirus present in the body or in various body fluids, it would be possible to determine whether the presence of disease, the course of disease, and/or whether a particular therapeutic regimen aimed at ameliorating the rotavirus disease is effective.
  • the monoclonal antibodies of the invention may be used in vitro in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier.
  • the monoclonal antibodies in these immunoassays can be detectably labeled in various ways.
  • types of immunoassays which can utilize monoclonal antibodies of the invention are competitive and non-competitive immunoassays in either a direct or indirect format.
  • Examples of such immunoassays are the radioimmunoassay (RIA) and the sandwich (immunometric) assay.
  • Detection of the antigens using the monoclonal antibodies of the invention can be done utilizing immunoassays which are run in either the forward, reverse, or simultaneous modes, including immunohistochemical assays on physiological samples. Those of skill in the art will know, or can readily discern, other immunoassay formats without undue experimentation.
  • the monoclonal antibodies of the invention can be bound to many different carriers and used to detect the presence of rotavirus.
  • carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, agaroses and magnetite.
  • the nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding monoclonal antibodies, or will be able to ascertain such, using routine experimentation.
  • labels and methods of labeling There are many different labels and methods of labeling known to those of ordinary skill in the art. Examples of the types of labels which can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, and bio-luminescent compounds. Those of ordinary skill in the art will know of other suitable labels for binding to the monoclonal antibodies of the invention, or will be able to ascertain such, using routine experimentation. Furthermore, the binding of these labels to the monoclonal antibodies of the invention can be done using standard techniques common to those of ordinary skill in the art. [0096] For purposes of the invention, human rotavirus may be detected by the monoclonal antibodies of the invention when present in biological fluids and tissues.
  • a sample containing a detectable amount of rotavirus can be used.
  • a sample can be a liquid such as urine, saliva, cerebrospinal fluid, blood, serum and the like, or a solid or semi-solid such as tissues, feces, and the like, or, alternatively, a solid tissue such as those commonly used in histological diagnosis.
  • Another labeling technique which may result in greater sensitivity consists of coupling the antibodies to low molecular weight haptens. These haptens can then be specifically detected by means of a second reaction. For example, it is common to use haptens such as biotin, which reacts with avidin, or dinitrophenol, pyridoxal, or fluorescein, which can react with specific anti-hapten antibodies.
  • the antibody of the present invention can be provided in a kit, i.e., a packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay.
  • the kit will include substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides the detectable chromophore or fluorophore).
  • substrates and cofactors required by the enzyme e.g., a substrate precursor which provides the detectable chromophore or fluorophore
  • other additives may be included such as stabilizers, buffers (e.g., a block buffer or lysis buffer) and the like.
  • the relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay.
  • the reagents may be provided as dry powders, usually lyophilized, including excipients which on dissolution will provide a reagent solution having the appropriate concentration.
  • the invention also provides isolated nucleic acids encoding the human anti-rotavirus antibodies, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the antibody.
  • Exemplary polynucleotides encode the heavy or light chain variable region sequences set forth herein, e.g. SEQ ID NO:1-18.
  • Nucleic acids of interest may be at least about 80% identical to a sequence that encodes SEQ ID NO: 1-18, at least about 85%, at least about 90%, at least about 95%, at least about 99%, or identical.
  • a contiguous nucleotide sequence is at least about 20 nt., at least about 25 nt, at least about 50 nt., at least about 75 nt, at least about 100 nt, and up to the complete coding sequence may be used.
  • Such contiguous sequences may encode a CDR sequence, or may encode a complete variable region. As is known in the art, a variable region sequence may be fused to any appropriate constant region sequence.
  • the nucleic acid encoding it is inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • DNA encoding the monoclonal antibody is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Many vectors are available.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the anti-rotavirus antibody of this invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous or homologous polypeptide, which include a signal sequence or other polypeptide having a specific cleavage site at the N- terminus of the mature protein or polypeptide, an immunoglobulin constant region sequence, and the like.
  • a heterologous signal sequence selected preferably may be one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
  • the signal sequence is substituted by a prokaryotic signal sequence selected.
  • An "isolated" nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the antibody nucleic acid.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the antibody where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase.
  • enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • the expressions "cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • Suitable host cells for cloning or expressing the DNA are the prokaryote, yeast, or higher eukaryote cells.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR(CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse Sertoli cells (TM4, Mather, Biol. Reprod.
  • monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TR1 cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1.982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • Host cells are transformed with the above-described expression or cloning vectors for anti-rotavirus antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the provided human antibody variable regions and/or CDR regions are used in a screening method to select for antibodies optimized for affinity, specificity, and the like.
  • random or directed mutagenesis is utilized to generate changes in the amino acid structure of the variable region or regions, where such variable regions will initially comprise one or more of the provided CDR sequences, e.g. a framework variable region comprising CDR1 , CDR2, CDR3 from the heavy and light chain sequences provided herein.
  • variable region sequences which are optionally combined with a second variable region sequence, i.e. V H VL, with constant regions, as a fusion protein to provide for display, etc., as known in the art.
  • Methods for selection of antibodies with optimized specificity, affinity, etc. are known and practiced in the art, e.g. including methods described by Presta (2006) Adv Drug Deliv Rev. 58(5-6) :640-56; Levin and Weiss (2006) Mol Biosyst. 2(1):49-57; Rothe et al. (2006) Expert Opin Biol Ther. 6(2):177-87; Ladner et al. (2001) Curr Opin Biotechnol.
  • Such screening methods may involve mutagenizing a variable region sequence comprising one or more CDR sequences set forth herein; expressing the mutagenized sequence to provide a polypeptide product; contacting the polypeptide with an rotavirus antigen; identifying those polypeptide having the desired antigen affinity or specificity.
  • the antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography is the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody.
  • Protein A can be used to purify antibodies that are based on human ⁇ 1 , ⁇ 2, or ⁇ 4 heavy chains (Lindmark et al., J. Immunol. Meth. 62:1-13 (1983)). Protein G is recommended for human ⁇ 3 (Guss et al., EMBO J. 5:15671575 (1986)).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH 3 domain, the Bakerbond ABXTM resin (J. T. Baker, Phillipsburg, N.J.) is useful for purification.
  • the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g., from about 0-0.25M salt).
  • the antibody formulations of the present invention may be used to treat the various rotavirus associated diseases as described herein.
  • the recipient is at a high risk of infection.
  • the antibody formulation is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the antibody formulation is suitably administered by pulse infusion, particularly with declining doses of the antibody.
  • the appropriate dosage of antibody will depend on the type of disease to be treated, the severity and course of the disease, whether the antibody is administered for preventive purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • an article of manufacture containing materials useful for the treatment of the disorders described above comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is one or more antibodies in a formulation of the invention as described above.
  • the label on, or associated with, the container indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Therapeutic formulations comprising one or more antibodies of the invention are prepared for storage by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • the antibody composition will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the "therapeutically effective amount" of the antibody to be administered will be governed by such considerations, and is the minimum amount necessary to reduce virus titer in an infected individual.
  • the therapeutic dose may be at least about 0.01 ⁇ g/kg body weight, at least about 0.05 ⁇ g/kg body weight; at least about 0.1 ⁇ g/kg body weight, at least about 0.5 ⁇ g/kg body weight, at least about 1 ⁇ g/kg body weight, at least about 2.5 ⁇ g/kg body weight, at least about 5 ⁇ g/kg body weight, and not more than about 100 ⁇ g/kg body weight. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, e.g. in the use of antibody fragments, or in the use of antibody conjugates.
  • the dosage may also be varied for localized administration, or for systemic administration, e.g. i.m., i.p., i.v., and the like.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat rotavirus infection. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active compound preferably those with complementary activities that do not adversely affect each other.
  • it may be desirable to further provide an antiviral agent.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom at least to some extent) of a disease state, e.g. to reduce virus titer in an infected individual.
  • the pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of subject being treated, subject-dependent characteristics under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered.
  • Oral administration can be accomplished using pharmaceutical compositions containing an agent of interest formulated as tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • Such oral compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets which can be coated or uncoated, can be formulated to contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients, e.g., inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, e.g., starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. Where a coating is used, the coating delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • non-toxic pharmaceutically acceptable excipients e.g., inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate
  • granulating and disintegrating agents for example, corn starch, or algin
  • the formulation is an aqueous suspension
  • such can contain the active agent in a mixture with a suitable excipient(s).
  • excipients can be, as appropriate, suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia); dispersing or wetting agents; preservatives; coloring agents; and/or flavoring agents.
  • Suppositories e.g., for rectal administration of agents, can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter and polyethylene glycols.
  • Dosage levels can be readily determined by the ordinarily skilled clinician, and can be modified as required, e.g., as required to modify a subject's response to therapy. In general dosage levels are on the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day. The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • Peptide Vaccine Compositions [00129] The application discloses herein a method of inducing an immune response against a peptide corresponding to an epitope recognized by an antibody disclosed herein, including without limitation specific epitopes of the VP4, VP5*, or VP7, where the epitope is of sufficient length to provide for binding specificity substantially similar to the specificity of binding to the native protein, e.g. a peptide of at least 20 amino acids, at least 30 amino acids, at least 40 amino acids, at least 50 amino acids, at least 100 amino acids, at least 150 amino acids, at least 200 amino acids up to the full length of the protein, where the peptide may be a contiguous or non-contiguous sequence of an rotavirus protein.
  • a basis for heterotypic neutralizing reactivity to RV in humans at the individual immunoglobulin (Ig) molecule level is identified.
  • a method of defining such activity comprising the steps of sorting single cells of intestinal RV-specific lgA + antibody secreting cells, by contacting the cells with triple-layered RV particles conjugated to a detectable label, e.g. a fluorochrome suitable for sorting by flow cytometry.
  • the immunoglobulin coding polynucleotides from such sorted cells are sequenced with an identifying barcode.
  • the antibodies thus identified by sequences are tested for activity in RV neutralization in vitro against two or more different RV serotypes, where antibodies that neutralize multiple serotypes are defined as heterotypic antibodies.
  • the methods are useful in providing detailed analysis of thre ability of an immunogen, e.g. a vaccine, to elicit a protective heterotypic response.
  • Humans can circumvent the serotypic diversity of naturally circulating RV strains by expressing individual VP4 epitope-specific Ig molecules that mediate heterotypic neutralization. Characterization of the structural targets of these mAbs, and determination of the extent to which they arise following primary RV infection of children provide the basis for designing more effective RV vaccines.
  • Antigenic compositions comprise all or a portion of a rotavirus protein in which specific highly immunodominant residues are masked or deleted, so as to generate an immune response to residues that are less immunodominant, but which are essential for virus function and therefore are less likely to be altered in virus escape mutation and selection.
  • antigenic compositions providing epitopes for heterotypic neutralizing antibodies are provided, which can be formulated alone or in combination with conventional vaccines.
  • Antigens may comprise, without limitation, VP5* proteins, alone or in combination with an adjuvant. These antigens find use in screening assays, generation of monoclonal antibodies, and in vaccines.
  • Such formulations may comprise, without limitation, live attenuated formulation containing known heterotypic neutralizing epitopes (and excluding known homotypic neutralizing epitopes); and/or epitope immunogens with known heterotypic neutralizing epitopes or overlapping neutralizing epitopes.
  • novel vaccines/immunogens could be used in combination with current formulations, for example in a prime boost strategy to enhance immunity in children and infants who do not respond to the current, licensed vaccines or formulations alone.
  • the formulations of the invention may find particular benefit in providing improved protective immunity in regions of the world with the highest RV disease burden and lowest vaccine efficacy observed in several clinical trials of the current licensed RV vaccines.
  • a modified rotavirus VP4, including a VP5* fragment, or VP7 polypeptide is provided, which provides for enhanced heterotypic immune responsiveness
  • a polynucleotide encoding such a modified rotavirus polypeptide is provided.
  • the polypeptide and/or the nucleic acid can be used in the formulation of a vaccine, e.g. a virus-like particle, a recombinant protein vaccine which can be formulated with an adjuvant, a vector vaccine, and the like.
  • a vaccine formulation comprising a polypeptide or a polynucleotide of the invention is provided.
  • portions of the rotavirus protein or live-attenuated whole virus are provided as an immunogen known to stimulate heterotypic protective immunity in humans as determined by epitope mapping studies using these mAbs. All or a portion of the rotavirus protein is provided as an antigen, where specific highly immunodominant residues are masked, so as to allow for the generation of an immune response to residues that are less immunodominant, but which are essential for virus function and therefore are less likely to be altered. These antigens find use in screening assays, generation of monoclonal antibodies, and in vaccines. Peptides for immunization may be conjugated to a carrier molecule prior to administration to a subject.
  • Peptides can be produced using techniques well known in the art. Such techniques include chemical and biochemical synthesis. Examples of techniques for chemical synthesis of peptides are provided in Vincent, in Peptide and Protein Drug Delivery, New York, N.Y., Dekker, 1990. Examples of techniques for biochemical synthesis involving the introduction of a nucleic acid into a cell and expression of nucleic acids are provided in Ausubel, Current Protocols in Molecular Biology, John Wiley, and Sambrook, et al in Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1989.
  • an immunologically effective amount of one or more immunogenic polypeptides which may be conjugated to a suitable carrier molecule, is administered to a patient by successive, spaced administrations of a vaccine, in a manner effective to result in an improvement in the patient's condition.
  • immunogenic polypeptides are coupled to one of a number of carrier molecules, known to those of skill in the art.
  • a carrier protein must be of sufficient size for the immune system of the subject to which it is administered to recognize its foreign nature and develop antibodies to it.
  • the carrier molecule is directly coupled to the immunogenic peptide.
  • the coupling reaction may require a free sulfhydryl group on the peptide.
  • an N-terminal cysteine residue is added to the peptide when the peptide is synthesized.
  • traditional succinimide chemistry is used to link the peptide to a carrier protein. Methods for preparing such peptidexarrier protein conjugates are generally known to those of skill in the art and reagents for such methods are commercially available (e.g., from Sigma Chemical Co.). Generally about 5-30 peptide molecules are conjugated per molecule of carrier protein.
  • Exemplary carrier molecules include proteins such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA), flagellin, influenza subunit proteins, tetanus toxoid (TT), diphtheria toxoid (DT), cholera toxoid (CT), a variety of bacterial heat shock proteins, glutathione reductase (GST), or natural proteins such as thyroglobulin, and the like.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • flagellin influenza subunit proteins
  • influenza subunit proteins tetanus toxoid
  • TT tetanus toxoid
  • DT diphtheria toxoid
  • CT cholera toxoid
  • GST glutathione reductase
  • natural proteins such as thyroglobulin, and the like.
  • the carrier molecule is a non-protein, such as Ficoll 70 or
  • VLPs virus capsid proteins that have the capability to self-assemble into virus-like particles
  • examples of VLPs used as peptide carriers are hepatitis B virus surface antigen and core antigen, hepatitis E virus particles, polyoma virus, and bovine papilloma virus.
  • a peptide vaccine composition may comprise single or multiple copies of the same or different immunogenic peptide, coupled to a selected carrier molecule.
  • the peptide vaccine composition may contain different immunogenic peptides with or without flanking sequences, combined sequentially into a polypeptide and coupled to the same carrier.
  • immunogenic peptides may be coupled individually as peptides to the same or a different carrier, and the resulting immunogenic peptide-carrier conjugates blended together to form a single composition, or administered individually at the same or different times.
  • peptide vaccine compositions are administered with a vehicle.
  • vehicle The purpose of the vehicle is to emulsify the vaccine preparation.
  • Numerous vehicles are known to those of skill in the art, and any vehicle which functions as an effective emulsifying agent finds utility in the present invention.
  • an immunological adjuvant may be included in the vaccine formulation.
  • Exemplary adjuvants known to those of skill in the art include water/oil emulsions, non-ionic copolymer adjuvants, e.g., CRL 1005 (Optivax; Vaxcel Inc., Norcross, Ga.), aluminum phosphate, aluminum hydroxide, aqueous suspensions of aluminum and magnesium hydroxides, bacterial endotoxins, polynucleotides, polyelectrolytes, lipophilic adjuvants and synthetic muramyl dipeptide (norMDP) analogs.
  • Suitable pharmaceutically acceptable carriers for use in an immunogenic proteinaceous composition of the invention are well known to those of skill in the art. Such carriers include, for example, phosphate buffered saline, or any physiologically compatible medium, suitable for introducing the vaccine into a subject.
  • Controlled release preparations may be achieved by the use of polymers to complex or absorb the peptides or antibodies. Controlled delivery may accomplished using macromolecules such as, polyesters, polyamino acids, polyvinyl pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate, the concentration of which can alter the rate of release of the peptide vaccine.
  • the peptides may be incorporated into polymeric particles composed of e.g., polyesters, polyamino acids, hydrogels, polylactic acid, or ethylene vinylacetate copolymers.
  • the peptide vaccine is entrapped in microcapsules, liposomes, albumin microspheres, microemulsions, nanoparticles, nanocapsules, or macroemulsions, using methods generally known to those of skill in the art.
  • the vaccine of the present invention can be administered to patient by different routes such as intravenous, intraperitoneal, subcutaneous, intramuscular, or orally.
  • a preferred route is intramuscular or oral.
  • Suitable dosing regimens are preferably determined taking into account factors well known in the art including age, weight, sex and medical condition of the subject; the route of administration; the desired effect; and the particular conjugate employed (e.g., the peptide, the peptide loading on the carrier, etc.).
  • the vaccine can be used in multi-dose vaccination formats.
  • a dose would consist of the range of to 1.0 mg total protein. In an embodiment of the present invention the range is 0.1 mg to 1.0 mg. However, one may prefer to adjust dosage based on the amount of peptide delivered. In either case these ranges are guidelines. More precise dosages should be determined by assessing the immunogenicity of the conjugate produced so that an immunologically effective dose is delivered.
  • An immunologically effective dose is one that stimulates the immune system of the patient to establish a level immunological memory sufficient to provide long term protection against disease caused by infection with rotavirus.
  • the conjugate is preferably formulated with an adjuvant.
  • the timing of doses depend upon factors well known in the art. After the initial administration one or more booster doses may subsequently be administered to maintain antibody titers. An example of a dosing regime would be a dose on day 1 , a second dose at or 2 months, a third dose at either 4, 6 or 12 months, and additional booster doses at distant times as needed.
  • the vaccine formulation is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. In addition, the vaccine formulation is suitably administered by pulse infusion, particularly with declining doses of the vaccine.
  • the appropriate dosage of vaccine will depend on the type of disease to be treated, the severity and course of the disease, whether the vaccine is administered for preventive purposes, previous therapy, the patient's clinical history and response to the vaccine, and the discretion of the attending physician.
  • the vaccine is suitably administered to the patient at one time or over a series of treatments.
  • an article of manufacture containing materials useful for the vaccination described above comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is one or more antibodies in a formulation of the invention as described above.
  • the label on, or associated with, the container indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Therapeutic formulations are prepared for storage by mixing the vaccine having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • the vaccine composition will be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • the "therapeutically effective amount" of the vaccine to be administered will be governed by clinical considerations, and is the minimum amount necessary to reduce virus titer in an infected individual.
  • An immunologically effective dose is one that stimulates the immune system of the patient to establish a level immunological memory sufficient to provide long term protection against disease caused by infection with rotavirus. More precise dosages should be determined by assessing the immunogenicity of the vaccine produced so that an immunologically effective dose is delivered.
  • the therapeutic dose may be at least about 0.01 ⁇ g/kg body weight, at least about 0.05 ⁇ g/kg body weight; at least about 0.1 ⁇ g/kg body weight, at least about 0.5 ⁇ g/kg body weight, at least about 1 ⁇ g/kg body weight, at least about 2.5 ⁇ g/kg body weight, at least about 5 ⁇ g/kg body weight, and not more than about 100 ⁇ g/kg body weight. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, e.g. in the use of vaccine fragments, or in the use of vaccine conjugates.
  • the dosage may also be varied for localized administration, or for systemic administration, e.g. i.m., i.p., i.v., and the like.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the formulation is an aqueous suspension
  • such can contain the active agent in a mixture with a suitable excipient(s).
  • excipients can be, as appropriate, suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia); dispersing or wetting agents; preservatives; coloring agents; and/or flavoring agents.
  • Suppositories e.g., for rectal administration of agents, can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter and polyethylene glycols.
  • the invention provides a means for classifying the immune response to peptide vaccine, e.g., 9 to 15 weeks after administration of the vaccine; by measuring the level of antibodies against the immunogenic peptide of the vaccine.
  • VP4- and VP7-specific antibodies mediate heterotypic immunity to rotavirus in humans.
  • VP5* the carboxy-terminal trypsin cleavage fragment of VP4
  • murine mAbs to VP8* the amino terminal trypsin cleavage fragment of VP4 are primarily serotype specific.
  • Rotateq contains five live, reassortant RVs each expressing serotypically distinct RV antigens, and Rotarix contains only a single RV strain.
  • An alternate hypothesis is that heterotypic immunity is mediated by an array of individual Ab molecules, each with restricted neutralizing specificity against a single RV serotype. Only one study has directly examined the serotypic specificity of human anti-VP4 and anti-VP7 mAbs.
  • TLP-Cy5 to B cells expressing surface VP4- or VP7-specific Ig was assessed by FACS in staining and blocking experiments using mouse hybridoma cells with previously characterized specificity for VP6 and for genotypically or serotypically distinct VP4s (P[8] or P[3]) and VP7s (G1 or G3).
  • CDC-9 TLP-Cy5 stained G1- and P[8]-specific hybridomas (mean fluorescence intensity (MFI) + SD) for G8 and P[8] of 9800 + 71 , 12801 + 282, respectively) but did not stain G3- or P[3]-specific hybridomas (Fig. 1 B, C).
  • Pre-incubation of G1- or P[8]-specific hybridomas with unlabeled CDC-9 TLPs reduced TLP-Cy5 binding to both hybridoma cell lines (G1 by 5- fold, P[8] by 7-fold).
  • TLP-Cy5 for these well-characterized murine hybridomas was established, we proceeded to stain isolated human intestinal B cells with the RV TLP-Cy5 preparation. Unstained control cells were used to identify TLP-specific lgA + and IgA " ASCs. Cold blocking with unlabeled TLPs reduced TLP-Cy5 staining on total intestinal B cells by 10 fold in lgA + ASCs and by 4 fold in IgA " ASCs (Fig. 1 D, E).
  • the CDC-9 TLP-Cy5 particles were then used to isolate RV-reactive B cells from proximal jejunum resections obtained from five adult patients undergoing bariatric surgery.
  • ASCs which include both plasmablasts and plasma cells, were identified by flow cytometry by gating on live, single CD3/14/16 " CD20'° CD27 hi CD38 hi cells.
  • lgA + ASCs were identified based on surface Ig expression.
  • TLP-binding lgA + ASCs were defined using gating based on unstained ASCs (Fig. 2A). The majority of intestinal ASCs in the five adult subjects were lgA + (median frequency 59.4%, range 53.4-82.5%).
  • Intestinal IgA " ASCs were detected at a median frequency of 40.6% (range 17.5-46.7%). Within the ASCs, most RV TLP-binding reactivity was detected among lgA + ASCs (median frequency 0.13%, range 0.09-1.30%). Some TLP-binding reactivity was also observed among IgA " ASCs (median frequency 0.01 %, range 0.00-0.04%) (Fig. 2B).
  • the median frequency of total lgA + ASCs as a proportion of total intestinal B cells was 35.2% (range 10.0-61.6%).
  • the median frequency of VP6-specific lgA + ASCs as a proportion of total lgA + ASCs was 0.16% (range 0.03-0.37%) (Fig. 2C).
  • a total of 821 paired IGHV and IGLV sequences were analyzed from the five donors; the median number of paired Ab sequences recovered per donor was 207 (range 82-227). Clonally related Abs were identified at an overall frequency of 29% of all paired sequences from all donors (9-60% in individual donors) (Fig. 3). The median number of clonal families per subject was 12 (range 8-16), with a range of 2-14 Abs per clonal family. The frequency of combinatorial IGVH-IGVL gene segment usage among all donors was analyzed. The majority (613) of VH-VL combinations were unique to individual donors. Sixteen (2.6%) IGVH-IGVL gene combinations were detected in two donors, and two (0.3%) sequences were detected in three donors. No combinations were detected in more than three donors (Fig. 4A).
  • RV- directed VP4- or VP7-specific mAbs were defined as those mAbs that bound to TLPs but not to purified DLPs or recombinant VP2/VP6 DL-VLPs.
  • VP6-specific mAbs were defined as those that bound specifically to purified DLPs and/or recombinant VP2/VP6 DL-VLPs whether or not they bound to TLPs.
  • Table 1 summarizes the protein specificities of the recombinant mAbs. Thirty-three of the 62 expressed mAbs bound to TLPs but not to DLPs, and hence were presumed to be either VP4- or VP7-specific.
  • mAbs displayed binding reactivity to cells infected with non- human RV strains: three to RRV, ST3, NCDV, and OSU; two to ST3; one to ST3, NCDV, and OSU; one to NCDV and OSU; one to ST3, RRV, and OSU; and one to ST3, RRV, and NCDV (Table 3).
  • the VP4/VP7-specific mAbs examined displayed varying but, in most cases, substantial degrees of heterotypic reactivity as measured by binding to cells infected with multiple RV serotypes.
  • six VP4/VP7-specific mAbs bound only to cells infected with Wa RV including three of the four VP7 mAbs (Table 3).
  • VP4-specific mAbs bound specifically to recombinant VP5*, the carboxy terminal stalk region of VP4. These five mAbs did not bind to recombinant VP8* (Table 1 , Table 3). VP5* binding was assessed by immunoprecipitation of in vitro translated VP5* as previously described. The binding site specificity of two VP4 specific mAbs could not be determined using these strategies (Table 1).
  • VP4- and VP7-specific intestinal-derived mAbs display neutralizing activity in vitro.
  • In vitro neutralization capacities of the VP4 and VP7 binding recombinant mAbs were assessed in assays using the Wa and CDC-9 RV strains (G 1 P[8]), three VP7 mono-reassortants including D x RRV (G1 P[3], DS1 x RRV (G2P[3]) and ST3 x RRV (G4P[3], and a set of serotypically distinct animal and human RV strains including DS1 (G2P[4]), RRV (G3P[3]), ST3 (G4P[6]), OSU (G5P[7]), NCDV (G6P[1 ]), UK (G6P[5]), 69M (G8P[10]), 1 16E (G9P[1 1 ]), WI61 (G9P[8]), and L26 (G12P[4
  • RV-reactive mAbs isolated from the five adult subjects neutralized one or more of these RV strains in vitro.
  • Three of the nine mAbs (mAb #27, #46 and #57) were VP7-specific as determined by their ability to neutralize the G 1 VP7 monoreassortant D x RRV but not the G3 parental RRV strain and by their specific immunostaining of Sf9 cells infected with BVs expressing Wa VP7.
  • Another six were VP4-specific as determined by specific binding assays to various forms of recombinant VP4.
  • homotypic neutralizing mAbs as mAbs for which the neutralization activity, defined by minimum neutralization concentration of the mAb, to a single serotype (G or P) was >10 fold higher than that to other serotypes.
  • Heterotypic mAbs were defined as those with minimum neutralization concentration within 10- fold for two or more distinct serotypes. Based on these criteria, three of the nine neutralizing Abs were homotypic: VP4 (VP5*)-specific mAb #33 (P[8]) and VP7-directed mAbs #27 (G1) and # 46 (G1).
  • mAb #49 demonstrated a low level of neutralizing activity with the highest minimum neutralizing concentration against a human RV strain at 39.1 ng/ml against Wa.
  • VP4- and VP7-specific intestinal mAbs display both homotypic and heterotypic neutralizing activity in vivo.
  • the ability of mAbs to protect against RV-induced diarrheal disease in vivo was examined using rhesus RRV (G3P[3]), a human RV VP7-RRV mono-reassortant D x RRV (G1 P[3]), the monoreassortant DS1 x SB1A (G4P4), and Wa RV (G1 P8) as challenge strains.
  • VP7-specific mAb #27 (G1 specific), when co-incubated with RRV (G3 serotype) or D x RRV (G1 serotype) and then administered orally at a dose of 10 6 PFU to 5-day-old suckling 129/Sv mice, prevented the G1 D x RRV-induced but not the G3 RRV-induced diarrheal disease (Fig. 5A).
  • VP7-directed mAb #57 which neutralized both G1 and G3 RV strains in vitro, protected against both RRV- and D x RRV-induced diarrhea at an efficacy of 100% (Fig. 5B).
  • mAb #41 which is directed at VP5* and neutralized both P[4] and P[8] RV strains in vitro, had a protective efficacy of 67% against the P[4] DS1 x SB1A monoreassortant and a 100% protective efficacy against the Wa P[8] (Fig. 5C).
  • the VP7-specific mAb #27 was able to inhibit RV-induced diarrhea in a VP7-serotype-specific manner, whereas VP7-specific mAbs #57 and VP5*-specific mAb #41 inhibited RV- induced diarrhea in a heterotypic manner.
  • RV vaccines like several other orally administered vaccines (e.g., cholera, typhoid, and polio vaccines), have less efficacy in developing countries than in developed countries. Multiple factors likely account for this effect including higher frequency of microbial pathogen co- infections, elevated levels of breast milk IgA or transplacental IgG specific to the vaccine at the time of vaccination, malnutrition, micronutrient deficiencies, the force of infection in less developed versus developed countries and the distinct microbiome of the vaccine recipients in less developed countries. Furthermore, the substantial serotypic diversity of circulating wild- type human RV strains is likely an impediment to the development of broadly effective RV vaccines in especially in less developed countries where RV serotypic diversity is greatest.
  • a barcode-based sequencing strategy was used to facilitate the efficient selection of natively paired, antigen-specific antibodies.
  • the strategy can accurately identify clonal expansions, if present in the Ab repertoire, as a proxy for antigen-activated and expanded B cells.
  • This approach has been shown to be highly effective in identifying clonally expanded and enriched antigen-specific plasmablasts with higher affinity and neutralizing capacity than singletons from the same patient, when applied to the analysis of peripheral antibody-secreting plasmablasts induced following recent vaccination, infection or other form of acute antigen exposure.
  • we use labeled antigen-specific bait to enrich for antigen- specificity in the steady state ASC repertoire from adult subjects who were unlikely to have an acute antigen-specific plasmablast response similar to those with recent vaccination or infection.
  • RV-binding Abs that did not have neutralizing activity appeared to have fewer somatic mutations in their VH genes compared to genes encoding RV neutralizing Abs and Abs not characterized in terms of binding specificity in this study.
  • VP5*- directed murine mAbs have demonstrated more cross-reactive serologic specificity.
  • the cross- reactivity of anti-VP5* Abs is consistent with the relative sequence conservation of this region and functional constraints on this portion of the molecule due to its role in membrane passage during cell entry. It is interesting to note that, like VP7, we have failed to identify any VP5* directed Abs that lacked the ability to restrict RV replication suggesting that most of the VP5* antigenic surface that is exposed on TLPs likely plays an important role in mediating viral infection.
  • the Ab-antigen co-evolution of heterotypic immunity to RV may have occurred in a manner similar to what is observed for broadly neutralizing human mAbs against the influenza membrane proximal HA stalk domain and HIV-1 envelope glycoprotein, both of which target receptor binding sites and membrane fusion machinery.
  • the precise atomic binding sites of the broadly heterotypic human VP5* mAbs described here as well as their mechanism of neutralization await additional studies; however, such Abs are unlikely to function by inhibiting viral binding but might be involved in restricting cell entry.
  • Prior studies using experimentally induced murine mAbs identified amino acid regions 248 to 474 as critical sites for the binding of heterotypic VP5*-directed heterotypic mAbs.
  • truncated VP8* subunit protein vaccine candidates containing most of the neutralizing epitopes expressed on VP8* have recently been shown to elicit RV-neutralizing Ab responses in animal models and to boost neutralizing Ab titers in RV-experienced adults when administered parenterally. It is surprising that none of the 22 individual anti-VP8* Abs isolated in our study had neutralizing activity in vitro in traditional cell culture assays, neutralization assays using human intestine derived organoids, or in passive protection challenge experiments in suckling mice.
  • VP8* fragments of VP4 of the major human RV serotypes interact with several distinct human histo-blood group antigens (HBGA), expressed on mucosal epithelial and other cell types. Genetic and developmental variation in HBGA expression may result in variable susceptibility to infection with different RV strains. P[8] and P[4] strains share reactivity with the common Lewis b (Le b ) and H type 1 antigens, whereas P[6] strains bind the H type 1 antigen only. Most VP8*-specific mAbs identified in this work bound to VP4 from both P[8] and P[4] strains (18/22).
  • Proximal jejunum tissue resections were obtained from adults undergoing bariatric surgery at the Stanford University Hospital in accordance with Stanford University IRB protocols (IRB Protocol 13813). Exclusion criteria included chronic viral infections or acute gastroenteritis at the time of surgery.
  • RV strains, propagation and preparation of TLPs, DLPs, and VLPs were grown in MA-104 cells (ATCC) in the presence of trypsin as described [88].
  • TLPs were purified from MA-104 cell lysates by genetron extraction, centrifugation through a sucrose cushion, and cesium chloride (CsCI) density gradient centrifugation as described [89, 90]. Purified TLPs were dialyzed to remove residual CsCI. DLPs were prepared by treating TLPs with 5 mM EDTA for 20 min at 37°C. VP2-eGFP/VP6 particles were prepared as previously described.
  • TLP preparation and labeling TLPs (CDC-9) were labeled with Cy5 as described [92] with some modifications. Varying molar ratios of Cy5 to TLP were tested to determine the TLP- Cy5 conjugate that yielded the highest signal to noise ratio in FACS staining with VP4- and VP7-specific hybridoma cells (data not shown). Briefly, TLPs (100 ⁇ g) were washed twice with 10 mM Hepes, pH 8.2, 5 mM CaCI 2 , 140 mM NaCI and labeled at 4: 1 molar ratio of Cy5 mono- reactive dye (GE Healthcare) to TLP at room temperature for 1 h with gentle agitation.
  • Cy5 mono- reactive dye GE Healthcare
  • TLP-Cy5 compared to unlabeled TLP was determined by electron microscopy as described.
  • Murine hybridomas VP6 (1 e1 1), VP4 P[8] (1 a10) or P[3] (7a12), VP7 G 1 (5e8) or G3 (159)) or enriched intestinal B cells were stained with TLP-Cy5 (2 ⁇ g) for 45 min on ice as previously described with modifications.
  • the concentration of TLP-Cy5 required per staining reaction was determined in titration experiments on VP4-, VP7-, and VP6-specific hybridomas.
  • Intestinal B cells were stained with LIVE/DEAD Fixable Acqua Dead Cell Stain Kit (Life Technologies) and a fluorescently-tagged Ab panel consisting of anti-CD3-PE Cy7 (clone: SKY), anti-CD14 PE Cy7 (clone: M5E2 ), anti-CD16-PE Cy7 (clone: 3G8), anti-CD20-APC H7 (clone: 2H7), anti-CD27-PE (clone: MT271), and anti-CD38 PerCP-Cy5.5 (clone: HIT2) all from Becton Dickinson and anti-lgA FITC (clone: IS1 1 -8E10, Miltenyi Biotec).
  • lgA + ASCs were identified by gating on live, single cells and CD3/14/16 " CD20 lo/" CD27 hi CD38 hi lgA + surface expression.
  • lgA + ASCs were bulk sorted using the Becton Dickinson FACS Aria III . The bulk- sorted population was then single-cell sorted into a 96-well PCR plate containing 10 mM Tris- HCI, pH 7.6, 2 mM dNTPs (New England Biolabs), 5 ⁇ oligo (dT) and 1 unit/ ⁇ Ribolock (Thermo Scientific). At least 200, 000 events were acquired per sample. Data were analyzed using Cytobank [94].
  • ELISPOT The frequencies of intestinal lgA + ASCs and VP6 + lgA + ASCs were determined by ELISPOT as described.
  • RT Reverse transcription
  • PCR with well-ID and plate-ID oligonucleotide barcode adaptors was performed as described [45]. Briefly, 6 mM MgCI 2 with Ribolock, Superscript III (Life Technologies), and 1 ⁇ of the appropriate well-ID oligonucleotide barcode were added to the sorted ASCs in individual wells of 96-well plates and RT was performed at 42 °C for 120 min. RT products from each plate were pooled.
  • PCR1 was performed with forward (FW) primers containing a 5' plate-ID barcode oligonucleotide and a 454 titanium adaptor, and with reverse primers specific for mRNAs encoding the Ig alpha, kappa, and lambda chains.
  • PCR2 was performed using FW primers with a 5' 454 titanium adaptor and reverse GSP with a 3' plate-ID barcode oligonucleotide and a 454 titanium adaptor. Amplified DNAs were pooled, purified with Ampure XP beads (Beckman Coulter) and sent to Roche for 454 sequencing.
  • V(D)J sequences were inserted into expression vectors containing interleukin-2 leader sequence (pFUSEss-CHIg-hG1 (lgG1), pFUSE2ss-CLIg-hK (IgK), pFUSE2ss-CLIg-hl_2 (IgL)) (InvivoGen). Supernatants were harvested after 5 days and assayed for IgG or IgA expression.
  • ELISAs The quantity of total IgG or IgA was assessed in transfection supernatants using the Human IgG or IgA ELISA kit (Zeptomatrix) and by fitting the standard curve to the 4 parameter logistic nonlinear regression model using Softmax Pro 6.5 (Molecular Devices). To determine binding reactivity to RV proteins, immunoplates (Thermo Fisher) were coated with TLPs (CDC-9), VLPs VP2-eGFP/VP6, bacterially expressed VP8* conjugated to tetanus toxoid [84] (a gift from PATH, Seattle, WA), or VP5* (1 ⁇ g/ml) overnight at 4 °C. VP5* was produced via in vitro transcription and translation as described.
  • Immunostaining was performed as previously described [50]. Recombinant BVs expressing VP7 (G1) or VP4 (Ku, DS-1 , 1076) were used to infect Sf9 cells at a multiplicity of infection of 0.1. Infected Sf9 cells were fixed with 10% formalin (Sigma) for 30 min at room temperature, and permeabilized with 1 % Triton X-100 (Sigma) in TNC (10 mM Tris, 100 mM NaCI, 1 mM CaCI 2 , pH 7.4) for 2 min at room temperature as previously described [50]. mAbs were serially diluted and incubated for 1 h at 37 °C.
  • mAbs that bound to specific BV- infected Sf9 cells were detected with HRP-labeled goat anti-human IgG or IgA (KPL), followed by incubation with 3-amino-9-ethyl-carbazole (AEC) (Vector Laboratories). The endpoint immunostaining concentration was assigned as the highest dilution at which cell staining could be detected using an inverted microscope.
  • MA104 cells were infected with specific RVs strains as indicated. Cells were fixed and permeabilized.
  • mAbs were used to stain intracellular RVs and binding reactivity was detected using HRP-conjugated goat anti-human IgG or IgA as described. All samples were run in duplicate and each assay was repeated twice.
  • MA104 cells were infected with human RV Wa strain at multiplicity of infection of 3. At 16 h post infection, total RNA was isolated using the RNeasy Mini Kit (Qiagen) according to manufacturer's instructions. cDNA was prepared from the isolated RNA using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems). VP4, VP5*, and VP8* coding sequences were amplified using Phusion polymerase (New England Biolabs) and the primers listed in Table 4. Amplified sequences were cloned into pCMV6-XL6 vector (Origene) containing SP6 promoter using Kpnl and Hindlll restriction sites.
  • VP4, VP5*, and VP8* proteins were translated in vitro using TNT®Quick Coupled Transcription/Translation Systems (Promega) with rabbit reticulocyte lysate and SP6 polymerase.
  • the translated proteins or whole virus particles were mixed with human anti-RV mAbs and incubated overnight at 4 °C with continuous mixing.
  • the protein-mAb complexes were then incubated for 1 hour at room temperature with PureProteome Protein A/G magnetic beads (Thermo Scientific) and precipitated using a magnetic field.
  • the immune-complexes were resolved in denaturing SDS-PAGE and immunoblotted to PVDF membrane.
  • the membranes were probed with anti-VP5* IgG (clone: HS-2). Immunoprecipitated proteins were visualized using ECL Plus western blotting substrate (Thermo Scientific).
  • Virus neutralization assays were performed as described
  • mAbs 5 ⁇ g/ml were serially diluted, and the dilutions were mixed with the following RV strains: Wa, CDC-9, D x RRV, DS1 , DS1 x RRV, RRV, ST3, ST3 x RRV, OSU, NCDV, UK, W161 , L26, 69M, 1 16E and 321 for 1 h at 37 °C.
  • the mAb-virus mixture was transferred to MA- 104 cell monolayers in a 96-well plate and incubated for 1 h at 37 °C in 5% C0 2 .
  • the antibody and virus mixture was removed, and cells were washed twice and incubated overnight at 37 °C with 100 ⁇ of M 199 media without serum or trypsin.
  • the cells were fixed with 10% formalin for 30 min and permeabilized with 1 % Triton X-100 for 2 min.
  • polyclonal rabbit anti-RV IgG was added to the plate for 2 h at 37 °C.
  • the plate was washed and HRP-conjugated goat anti-rabbit IgG ( ⁇ chain specific) (Sigma-Aldrich) was added. After 1 h incubation at 37 °C, a color reaction was detected with the AEC substrate.
  • the neutralization activity was defined as the highest dilution at which virus-positive foci were reduced by at least 50% compared to the controls untreated with mAb and expressed as minimum neutralization concentration (ng/ml). All samples were run in duplicate, and each assay was repeated twice.
  • the organoid cultures were switched to differentiation media comprised of growth media without Wnt3A, BS202190, or nicotinamide and with a 50% reduction of Noggin and R-spondin.
  • Human mAb #41 (5 ⁇ g/ml) or mixtures of VP8*-specific mAbs (mAb #4, #9, #16, #18, and #20 at 5 pg/ml) were incubated with Wa (10 s PFU) for 1 h at 37 °C.
  • the organoids were treated with TrypLE (Gibco) and co-incubated with Wa-mAb mixtures for 1 h at 37 °C.
  • mice were originally purchased from Taconic Biosciences. Sucking mice were bred in the VA Palo Alto Health Care System Veterinary Medical Unit. RVs were incubated with RV neutralizing human mAbs (5 ⁇ g/ml) for 1 h at 37 °C, and the RV-mAb mixture was then used to orally gavage 5 day old 129/Sv suckling mice.
  • Human anti-VP7 mAbs (mAb #27 and mAb #57) were mixed with RRV or D x RRV and human anti-VP4 mAb (mAb #41) was mixed with Wa or DS1 x SB1 A. Six to 1 1 mice were included per group.
  • the RV dose for each inoculum was 10 6 PFU. Mice were monitored for 4 days for diarrheal disease. All experiments were conducted in accordance with Stanford University and the VA Palo Alto Health Care System guidelines. mAb protective efficacy was calculated as: diarrhea rate of RV-infected control mice minus diarrhea rate of RV infected and mAb treated mice divided by the diarrhea rate of RV-infected control mice.
  • mAb ID corresponds to the identification numbers used in, for example, Tables 1-3. As indicated in the Tables, each of monoclonal antibodies 2, 27, 30, 33, 41 , 46, 47, 49 and 57 show neutralizing activity in vitro. Monoclonal antibodies 27, 41 and 57 have demonstrated in vivo neutralization activity. Monoclonal antibodies 2, 30, 33, 41 and 49 bind to VP5*; and 2, 30, 41 and 49 are heterotypic.
  • mAb ID: 2 comprises the heavy chain variable region (SEQ ID NO: 19) IGHEVQLVESGGGLVKPGGSLRLSCKASGLIVSDAWMSvWRQSPGKGLEVWGRIKSEINGGTI DYAAPVKGRFTILRDDSKNTLYLQINSLKTEDTAVYYCTTRLLFSPWGQGTLVTVSS, and the light chain variable region (SEQ ID NO:20)
  • mAb ID: 27 comprises the heavy chain variable region (SEQ ID NO:21)
  • mAb ID: 30 comprises the heavy chain variable region (SEQ ID NO:23)
  • mAb ID: 33 comprises the heavy chain variable region (SEQ ID NO:25) I G H DVQLVESGG G LVQ PG GPSRLSCSASRFTFSNYAMYVWRQAPGKGLEYVSSISSDGGSTY YAESVKGRFTISRDNSKNTLYLQMRSLRAEDAAVYYCVTDVLRLPYSTGWSPGDFIYWGQGT LVTVSS and the light chain variable region (SEQ ID NO:26) DIQMTQSPSILYASVGDRVTITCRASQSVSSWLAWYQQKPGKVPKLLIYQASTLENGVPSRFS GSGSGTEFILTISSLQPDDFATYYCQHYNVLWTFGQGTKVEI
  • mAb ID: 41 comprises the heavy chain variable region (SEQ ID NO:27) I G H EVQ LVESGGGPVQ PGGSLKLSCAASGFTFSNYEMYVWRQAPG KGLEVWSYISTSPAITY YADSVRGRFTISRDNAKSSLYLHMNSLRAEDTAVYYCATISHQQFSSGWNAWFDPWGQGTLV TVSS and the light chain variable region (SEQ ID NO:28) NFMLTQPHSVSESPGKTVTISCTGSSGSIASNYVQWYRQRPGSAPTTVIYENYQRPSGVPARF SGSIDRSSNSASLTISGLQTDDEADYYCQSYDNNNLVWFGGGTKLTVL
  • mAb ID: 46 comprises the heavy chain variable region (SEQ ID NO:29) QVQLQESGPGLVKPSETLSLTCTVSGGSINSYYWSWIRQSPGKGLEWIGYVFYSGITKYNPSL QSRVTISLDMGKNQFSLKLTSVNAADAAVYYCARNFPSYTPDWFFDLWGRGTLVTVSS and the light chain variable region (SEQ ID NO:30)
  • mAb ID: 47 comprises the heavy chain variable region (SEQ ID NO:31) QVQLQESGPGLVKPSETLSLTCSVSGGSISVYYWNWIRQSPGKGLEWIASMYYTGITNYNPSL KSRVTMSVDMSKNQFSLKLSSVTAADTAVYYCARTMGIDQNNRGWPPAGYYFGMDVWGQG TTVTVSS and the light chain variable region (SEQ ID NO:32) DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGNNYLDWYLQKPGQSPQLLIYLGSNRASGV PDRFSGSGSGTDFTLKISRVEAEDVGIYYCMQALEASLTFGGGTKVEIK
  • mAb ID: 49 comprises the heavy chain variable region (SEQ ID NO:33) DVQLVESGGGLVQPGGSVRLSCSASRFTFSNYAMYVWRQAPGKGLEYVSSISSDGGSTYYA ESVKGRFTISRDNSKNTLYLQMRSLRAEDAAVYYCVTDVLRLPYSTGWSPGDFIYWGQGTLVT VSS and the light chain variable region (SEQ ID NO:34) DIQMTQSPSILYASVGDRVTITCRASQSVSSWLAWYQQKPGKVPKLLIYQASTLENGVPSRFS GSGSGTEFILTISSLQPDDFATYYCQHYNVLWTFGQGTKVEIK
  • mAb ID: 57 comprises the heavy chain variable region (SEQ ID NO:35) QVQLVESGGGWQSGRSLRLSCAASGFTFRSYAMHVWRQAPGKGLEVWADLSLDGSHKYA DSVRGRFTISSDSSKNTVYLQMNSLRTEDTAIYYCARAAGIMVAGTFLTEFYFDYWGQGTLVT VSS and the light chain variable region (SEQ ID NO:36) QSVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNIKRPSGVPDR FSGSKSGTSASLAITGLQTEDEADYYCQSYDSSLSAYYVFGTGTRVTVL.
  • SEQ ID NO:35 the heavy chain variable region
  • VHH monovalent llama-derived antibody fragments

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Communicable Diseases (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des compositions et des méthodes relatives à des sérotypes de rotavirus et à des anticorps qui se lient au rotavirus humain, et à des modifications de ceux-ci, qui améliorent l'immunogénicité de la protéine du rotavirus pour le développement de vaccins en termes de génération d'une réponse immunitaire neutralisante. L'invention concerne également des méthodes d'utilisation des anticorps pour traiter une maladie médiée par le rotavirus chez un sujet.
PCT/US2016/041613 2015-07-08 2016-07-08 Anticorps hétérotypiques spécifiques contre le rotavirus humain WO2017008049A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/737,634 US20180155411A1 (en) 2015-07-08 2016-07-08 Heterotypic antibodies specific for human rotavirus

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562189901P 2015-07-08 2015-07-08
US62/189,901 2015-07-08

Publications (1)

Publication Number Publication Date
WO2017008049A1 true WO2017008049A1 (fr) 2017-01-12

Family

ID=57686076

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/041613 WO2017008049A1 (fr) 2015-07-08 2016-07-08 Anticorps hétérotypiques spécifiques contre le rotavirus humain

Country Status (2)

Country Link
US (1) US20180155411A1 (fr)
WO (1) WO2017008049A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023194913A1 (fr) * 2022-04-05 2023-10-12 Boehringer Ingelheim Vetmedica Gmbh Composition immunogène utile pour la vaccination contre les rotavirus

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230340082A1 (en) * 2020-07-10 2023-10-26 The Wistar Institute Of Anatomy And Biology Dna antibody constructs for use against rotavirus
CN116355079B (zh) * 2022-08-19 2023-12-26 上海迈科康生物科技有限公司 检测重组人轮状病毒vp8抗原(vp8 p[8])的单克隆抗体及其应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050031614A1 (en) * 2003-08-08 2005-02-10 Lorin Roskos Antibodies directed to parathyroid hormone (PTH) and uses thereof
WO2005049642A2 (fr) * 2003-11-21 2005-06-02 Institut Pasteur Genome des souches paris et lens de legionella pneumophila applications diagnostiques et epidemiologiques
US20110171316A1 (en) * 2008-05-29 2011-07-14 The Government Of The Us, As Represented By The Secretary, Department Of Health And Human Services Expression and assembly of human group c rotavirus-like particles and uses thereof
US20120058906A1 (en) * 2008-11-07 2012-03-08 Vaughn Smider Combinatorial antibody libraries and uses thereof
WO2014178820A1 (fr) * 2013-04-29 2014-11-06 Teva Pharmaceuticals Australia Pty Ltd. Anticorps anti-cd38 et fusions sur un interféron alpha-2b atténué

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050031614A1 (en) * 2003-08-08 2005-02-10 Lorin Roskos Antibodies directed to parathyroid hormone (PTH) and uses thereof
WO2005049642A2 (fr) * 2003-11-21 2005-06-02 Institut Pasteur Genome des souches paris et lens de legionella pneumophila applications diagnostiques et epidemiologiques
US20110171316A1 (en) * 2008-05-29 2011-07-14 The Government Of The Us, As Represented By The Secretary, Department Of Health And Human Services Expression and assembly of human group c rotavirus-like particles and uses thereof
US20120058906A1 (en) * 2008-11-07 2012-03-08 Vaughn Smider Combinatorial antibody libraries and uses thereof
WO2014178820A1 (fr) * 2013-04-29 2014-11-06 Teva Pharmaceuticals Australia Pty Ltd. Anticorps anti-cd38 et fusions sur un interféron alpha-2b atténué

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
"UniProtKB_A8TIEO, TRAP dicarboxylate transporter, DctP subunit. UniProtKB accession number: A8TIE0.", SEQUENCE LAST MODIFIED, 15 January 2008 (2008-01-15), Retrieved from the Internet <URL:http://www.uniprot.org/uniprot/A8TIEO> [retrieved on 20160825] *
HIGO-MORIGUCHI ET AL.: "Isolation of Human Monoclonal Antibodies That Neutralize Human Rotavirus.", J VIROL, vol. 78, no. 7, 2004, pages 3325 - 32, XP002367859 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023194913A1 (fr) * 2022-04-05 2023-10-12 Boehringer Ingelheim Vetmedica Gmbh Composition immunogène utile pour la vaccination contre les rotavirus

Also Published As

Publication number Publication date
US20180155411A1 (en) 2018-06-07

Similar Documents

Publication Publication Date Title
TWI803523B (zh) Tigit抗體、其抗原結合片段及醫藥用途
US20190256579A1 (en) Middle east respiratory syndrome coronavirus immunogens, antibodies, and their use
ES2789348T3 (es) Anticuerpos neutralizantes para GP120 y sus usos
TWI528974B (zh) 用於增加肌肉生長之組合物及方法
TWI758558B (zh) Cd96抗體、其抗原結合片段及醫藥用途
CN111744007B (zh) 一种抗tigit抗体药物组合物及其用途
CN113728003A (zh) 重组多克隆蛋白及其使用方法
US11834494B2 (en) Antibodies to zika virus and methods of use thereof
MX2015001600A (es) Anticuerpos anti-virus del dengue y usos de los mismos.
CN111417651B (zh) 多瘤病毒中和抗体
BRPI0911699B1 (pt) vetor, célula, composição farmacêutica, uso de um anticorpo, ou fragmento de ligação a antígeno deste
CN115768790A (zh) 针对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的人单克隆抗体
BR112019013468A2 (pt) anticorpos anti-hiv-1 amplamente neutralizante e métodos de uso dos mesmos
EP3220947B1 (fr) Anticorps à puissante activité de neutralisation du virus de la rage et d&#39;autres lyssavirus et leurs utilisations
US10081675B2 (en) Antibodies targeting M-CSF
US20240117011A1 (en) Antibodies targeting the spike protein of coronaviruses
US20220265806A1 (en) Antibodies or antibody-fragments thereof targeting alphaviruses, and compositions and methods comprising same
JP2024509054A (ja) 抗体
JP2022541652A (ja) エンテロウイルスd68に対するヒトモノクローナル抗体
US20200317753A1 (en) Humanized Antibodies Against Enterovirus 71
JP2022507836A (ja) 新規の抗ジカウイルス抗体及びその使用
US20180155411A1 (en) Heterotypic antibodies specific for human rotavirus
US20230085439A1 (en) Antibodies that bind human metapneumovirus fusion protein and their use
US20230272048A1 (en) Hiv-1 antibodies
US20130084301A1 (en) Cluster of Neutralizing Antibodies to Hepatitis C Virus

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16822065

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15737634

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16822065

Country of ref document: EP

Kind code of ref document: A1